WO2001009193A1 - Chimeric polypeptides, method for production and uses thereof - Google Patents

Chimeric polypeptides, method for production and uses thereof Download PDF

Info

Publication number
WO2001009193A1
WO2001009193A1 PCT/EP2000/007378 EP0007378W WO0109193A1 WO 2001009193 A1 WO2001009193 A1 WO 2001009193A1 EP 0007378 W EP0007378 W EP 0007378W WO 0109193 A1 WO0109193 A1 WO 0109193A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
biologically active
cysteines
amino acids
protein
Prior art date
Application number
PCT/EP2000/007378
Other languages
English (en)
French (fr)
Inventor
Hauke Lilie
Susanne Richter
Rainer Rudolph
Kay-Gunnar Stubenrauch
Original Assignee
F. Hoffmann-La Roche Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by F. Hoffmann-La Roche Ag filed Critical F. Hoffmann-La Roche Ag
Priority to DE60033405T priority Critical patent/DE60033405T2/de
Priority to MXPA02001139A priority patent/MXPA02001139A/es
Priority to BR0012926-7A priority patent/BR0012926A/pt
Priority to AU61610/00A priority patent/AU768645B2/en
Priority to DK00948017T priority patent/DK1206495T3/da
Priority to CA002380569A priority patent/CA2380569C/en
Priority to EP00948017A priority patent/EP1206495B1/en
Priority to JP2001513999A priority patent/JP4011914B2/ja
Publication of WO2001009193A1 publication Critical patent/WO2001009193A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/624Disulfide-stabilized antibody (dsFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/22011Polyomaviridae, e.g. polyoma, SV40, JC
    • C12N2710/22022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/22011Polyomaviridae, e.g. polyoma, SV40, JC
    • C12N2710/22023Virus like particles [VLP]

Definitions

  • the invention concerns chimeric polypeptides which are chemically linked via cysteine-based disulfide bridges, methods for production and uses thereof.
  • Bifunctional proteins are used particularly preferably in immunodiagnostics and immunotherapy.
  • advantage is taken of the specific binding of an antibody, or of an antibody fragment, to its antigen in order to direct a protein having a different biological function towards this specific antigen.
  • a bispecific antibody whose antigens are located, on the one hand, on tumor cells, and on the other, on macrophages can be utilized for directing killer cells towards a tumor (Bohlen, H. et al., Blood 82 (1993) 1803-1812).
  • bispecific antibodies can be produced by fusing two hybridoma cells, which produce the respective monospecific antibodies, to form quadroma cells (Milstein, C. and Cuello, A. C, Nature 305 ( 1983) 537- 540). Besides the two original antibodies these cells also produce bispecific antibodies. This method of obtaining bispecific proteins is, however, limited exclusively to antibodies. Furthermore, only 15% of the expressed antibodies exhibit the desired bispecificity. These antibodies have to be isolated from the mixture by means of labor-intensive purification methods.
  • a markedly more efficient method of producing bispecific antibodies and other bispecific proteins is based on the chemical cross-linking of two proteins having the desired properties (Fanger, M.W. et al., Crit. Rev. Immunol. 12 ( 1992) 101-24).
  • This cross-linking is accomplished by means of bifunctional linker molecules which react with amino groups of the proteins or cysteine residues.
  • cysteine residues of the one protein may be activated by 5,5'-dithiobis-(2- nitrobenzoic acid) (DTNB), and the addition of the second protein which contains cysteine residues in reduced form causes the formation of disulfides and, thus, covalent coupling of the two proteins.
  • DTNB 5,5'-dithiobis-(2- nitrobenzoic acid)
  • the most frequently applied method of producing bifunctional proteins is the formation of fusion proteins at the genetic level.
  • the 5' end of the cDNA of a protein is linked with the 3' end of the gene of another protein by genetic engineering methods, while the reading frame is retained, and the construct is expressed recombinantly in prokaryotes or eukaryotes.
  • an antibody fragment which was directed against a tumor was fused with a bacterial toxin (Brinkmann, U. et al., Proc. Natl. Acad. Sci. USA 88 (1991) 8616- 8620).
  • This immunotoxin is capable of specifically killing tumor cells.
  • diabodies represent another form of fusion proteins (Holliger, P. et al., Proc. Natl. Acad. Sci. USA 90 ( 1993) 6444-6448).
  • the diabodies consist of two Fv fragments having different specificities. Unlike scFv fragments in which the two variable domains of an antibody are connected to one another via a linker, in the case of diabodies the VL domain of the one antibody is fused with the VH domain of a second antibody.
  • the structure of the linker used for this purpose is such that the linker prevents an intramolecular association of the two domains, and instead of this, an intermolecular association of two of these constructs occurs, resulting in the formation of a bifunctional diabody.
  • the invention comprises a chimeric polypeptide consisting of a first and a second polypeptide chain being chemically linked via 1 to 3 cysteine-based disulfide bridges, characterized in that said first polypeptide chain consists of 1 to 3 cysteines and 4 to 12 basic amino acids, preferably selected from the group consisting of arginine, lysine and ornithine, and said second polypeptide chain consists of 1 to 3 cysteines and 4 to 12 acidic amino acids, preferably selected from the group consisting of glutamate and aspartate, whereby each of said polypeptide chains is linked at its C- and/or N- terminus to a biologically active compound.
  • said biologically active compounds differ from each other by their chemical structure. It is further preferred that said compound is an antibody, an antibody fragment or an enzyme.
  • the invention further comprises a method for the production of a chimeric polypeptide being chemically linked via 1 to 3 cysteine-based disulfide bridges and consisting of a first and a second polypeptide chain, wherein said first polypeptide chain consists of 1 to 3 cysteines and 4 to 12 basic amino acids, preferably selected from the group consisting of arginine, lysine and ornithine, and said second polypeptide chain consists of 1 to 3 cysteines and 4 to 12 acidic amino acids, preferably selected from the group consisting of glutamate and aspartate, whereby each of said polypeptide chains is fused at its C- and/or N-terminus to a biologically active compound, characterized in that two nucleic acids encoding said two polypeptide chains, which are each linked to a biologically active polypeptide, are expressed in a prokaryotic or eukaryotic host cell simultaneously or separately, the polypeptides are recovered from the host cell or the supernatant, treated with an oxid
  • the invention is based upon the fact that a polypeptide chain consisting of 4 to 12 basic amino acids specifically interacts in an aqueous solution at low ionic strength with a polypeptide chain consisting of 4 to 12 acidic amino acids. If both polypeptides additionally contain cysteines, in a subsequent reaction, disulfide bridges can be specifically formed between the cysteines of both polypeptide chains under oxidizing, or even slightly reducing, conditions. Preferably, the distance between two cysteines in the polypeptide chain is more than one amino acid, preferably 3 to 6 amino acids.
  • the amount and distance of the cysteines of both polypeptides are preferably identical and that if the cysteines of both polypeptides are positioned opposite each other, then acidic amino acids of the one strand and the basic amino acids of the other strand are in each case also positioned opposite.
  • Identical distance of the cysteines means that in both strands the same amount of amino acids other than cysteine is positioned between said cysteines.
  • the first and second polypeptide chains are designed in such a way that the position of the cysteines and of the basic and acidic amino acids enable an optimized ionic interaction between the acidic and basic amino acids and coupling via one or more disulfide bridges, in a preselected directed form. In this way it is possible to bring the biologically active compounds into a preselected spatial position with respect to one another.
  • the biologically active compounds can be brought into a very close spatial position relative to one another, if they are both linked at the C-terminus (or are both linked at the N- terminus) of the polypeptide chains. If one of the biologically active compounds is linked at the C-terminus and the other is linked at the N-terminus of the polypeptide chains, their spatial distance from one another is much greater. In such case, the dimerized first and second polypeptide chains essentially act as a linear linker between two biologically active compounds, preferably between two polypeptides.
  • An analogous procedure can be adopted when four biologically active compounds are to be brought into a spatial position relative to one another.
  • crizomeric polypeptide is meant a polypeptide that consists of a first and a second polypeptide chain, which chains differ chemically and are composed in such a way that they bind to one another as a result of ionic interaction via a plurality of amino acids carrying different charges and that they are additionally covalently bound to one another via disulfide bridges of cysteines.
  • the polyionic interactions between the acidic and basic amino acids in the polypeptide chains serve to bring the polypeptides into a preselected position relative to one another, which position makes it possible to easily form the disulfide bridges.
  • the purpose of the chimeric polypeptides is to couple the biologically active compounds to one another in a stable and predefined manner, avoiding undesired byproducts (e.g., homodimers, or chimeric products where the biologically active compounds are in an unfavorable position relative to one another).
  • the first and second polypeptide chains per se exhibit no substantial biological activity. They merely are auxiliary agents assisting in the coupling of the biologically active compounds, which, in their dimeric or multimeric form, are able to develop a preselected pharmaceutical effect.
  • the pK a values of the basic and acidic amino acids which are positioned on the chains in a position opposite to one another should differ as much as possible. Therefore, it is preferred that the pK a .value of the basic amino acid is about 10 or more whereas the pK a value of the acidic amino acid is about 4.5 or less.
  • the polypeptide chains consist of 1 to 3 cysteines and 4 to 12 additional amino acids, which are selected, for the first polypeptide chain, preferably from the group consisting of arginine, lysine and ornithine.
  • the second polypeptide chain consists of 1 to 3 cysteines and 4 to 12 acidic amino acids, preferably selected from the group consisting of glutamate and aspartate.
  • the polypeptide chain consists of 6 to 10 basic or acidic amino acids. It is further preferred that the polypeptide chains each contain a cysteine residue.
  • other acidic or basic amino acids or derivatives thereof can be used according to the invention as long as their pK a -values differ considerably (preferred difference about
  • Any desired biologically active compound can be used as the biologically active compound.
  • the term “biologically active compound or material” as used herein therefore refers to an organic molecule including a drug, a biological macromolecule such as a peptide, protein, carbohydrate (including monosaccharides, oligosaccharides, and polysaccharides), nucleoprotein, mucoprotein, lipoprotein, synthetic polypeptide or protein, or a small molecule linked to a protein, glycoprotein, steroid, nucleic acid (any form of DNA, including cDNA, or RNA, or a fragment thereof), nucleotide, nucleoside, oligonucleotides (including antisense oligonucleotides), gene, lipid, hormone, vitamin, including vitamin C and vitamin E, or a combination thereof, that causes a biological effect when administered in vivo to an animal, including but not limited to birds and mammals, including humans.
  • a biological macromolecule such as a peptide, protein, carbohydrate (including monosaccharides, oligosaccharides, and poly
  • the term broughtdrug refers to any substance used internally or externally as a medicine for the treatment, cure or prevention of a disease or disorder and includes but is not limited to immunosuppressants, antioxidants, anesthetics, chemotherapeutic agents, steroids (including retinoids), hormones, antibiotics, antivirals, antifungals, antiproliferatives, antihistamines, anticoagulants, antiphotoaging agents, melanotropic peptides, nonsteroidal and steroidal anti- inflammatory compounds, and radiation absorbers, including UV-absorbers.
  • the term facedavically active agent also includes agents such as insecticides, pesticides, fungicides, rodenticides, and plant nutrients and growth promoters.
  • Bio macromolecules preferably having a molecular weight in the range of two or three thousand to many millions are important regulators of physiological functions.
  • the size and tertiary structure of a biologically active macromolecule convey significant chemical information through highly specific interactions with receptors, enzymes, nucleic acids or other biological mediators interacting with it. Events as diverse as thrombosis, inflammation and immunologic responses are controlled, at least in part, by the three-dimensional topology of macromolecules.
  • the surface of the macromolecule is composed of geometrically distributed groups which impart ionic, hydrophobic, steric, electrostatic and hydrogen bonding character to the molecule and provide the molecular template for receptor binding.
  • Acid mucopolysaccharides also referred to as glycosaminoglycans (GAG) consist of recurring disaccharide units, each of which contains a derivative of an aminohexose, usually D-glucosamine or D-galactosamine. At least one of the two sugars in the recurring disaccharide unit of acid mucopolysaccharides contains an acidic group having a negative charge at pH 7, either a carboxylate or a sulfate group.
  • An important acid mucopolysaccharide is heparin, which is generated by certain types of cells that are especially abundant in the lining of arterial blood vessels. Heparin is a very powerful inhibitor of blood clotting and aids in preventing the formation of blood clots in circulating blood (Jackson, R. L., et al., Physiol. Reviews 71 (1991) 481-522).
  • GAG is a mediator of cellular processes (angiogenesis, nerve cell development, smooth muscle cell proliferation), gene expression and homeostasis. GAG interacts with DNA (Davidson, J. N., in “The biochemistry of the nucleic acids” Methuem, London, 1969).
  • Both DNA and GAG are linear polymers that have polyanionic charges that are essential for biological activity.
  • the rigidity of the DNA helix insures that the specifically sequenced nucleic acids are presented so as to obtain a desired biological interaction.
  • Proteins are the most abundant macromolecules in cells, making up over half their dry weight. Proteins and peptides are known to carry chemical information in their tertiary structures. A number of proteins occurring in nature are conjugated to other chemical groups. Examples are lipoproteins, glycoproteins, phosphorproteins, hemoproteins, flavoproteins and metalloproteins. Proteins have diverse biological functions.
  • Nonlimiting examples are transport proteins (e.g., hemoglobin and serum albumin), nutrient and storage proteins (for example, gliadin, ovalbumin, casein, and ferritin); contractile or motile proteins (e.g., actin, myosin, tubulin, and dynein); structural proteins (for example, keratin, fibroin, collagen, elastin, and proteoglycans); defense proteins (e.g., antibodies, immunoglobulins, fibrinogen, thrombin, botulinus toxin, diphtheria toxin, snake venom, and ricin); enzymes, and regulatory proteins (e.g., insulin, growth hormone, corticotropin and repressors).
  • transport proteins e.g., hemoglobin and serum albumin
  • nutrient and storage proteins for example, gliadin, ovalbumin, casein, and ferritin
  • contractile or motile proteins e.g., actin, myosin, tub
  • Hormones are classified as peptide hormones such as thyrotropin-releasing factor, corticotropin, vasopressin, insulin, and glucagon; amine hormones such as adrenaline and thyroxine; or steroid hormones such as cortisol, .beta.-estradiol, testosterone, and progesterone).
  • important hormones include, but are not limited to, adrenocorticotropin-releasing hormone, somatotropin releasing hormone, somatostatin, prolactin-releasing hormone, prolactin- inhibitory hormone, FSH- and LH-releasing hormone, vasopressin, and oxytocin.
  • Therapeutic biologically active compounds may also be selected from the general group consisting of anti-neoplastic agents, anti-infective agents, anti-depressives, antiviral agents, anti-nociceptive agents, anxiolytics and hormones.
  • anti-neoplastic agents useful in the compositions and methods of the present invention include methotrexate, taxol, tumor necrosis factor, chlorambucil, interleukins, bleomycin, etoposide, fluorouracil and vinblastine.
  • anti-viral agents useful in the compositions and methods of the present invention include dideoxyoytidine, zidovudine, acyclovir, interferons, dideoxyinosine and ganciclovir.
  • anxiolytics and sedatives useful in the compositions and methods of the present invention include benzodiazepines such as diazepam, barbiturates such as phenobarbital and other compounds such as buspirone and haloperidol.
  • hormones useful in the compositions and methods of the present invention include estradiol, prednisone, insulin, growth hormone, erythropoietin, and prostaglandins.
  • compositions and methods of the present invention including local anesthetics, vitamins, vaccines, wound healing stimulators, immunosuppressives, anti-emetics, anti-malarial agents, anti-fungal agents, anti-psychotics, anti-pyretics, coagulants, diuretics, calcium channel blockers, bronchodilatory agents, etc.
  • the biologically active compounds can be linked to the polypeptide chains according to the methods known in the art, for example by chemical coupling via reactive groups such as amino or carboxyl groups. Such methods are described, for example, in Mattson et al., Mol. Biol. Rep. 17 (1993) 167 - 183.
  • the biologically active compound is a polypeptide
  • chimeric polypeptides wherein the biologically active compounds are two different antibodies or antibody fragments (Fab, Fc, Fv fragments) or wherein one biologically active compound is an antibody or an antibody fragment and the other is a polypeptide having enzymatic activity like kinases, phosphatases, RNases, toxins or specific binding activities such as, for example, transcription factors.
  • the first biologically active compound is a ligand for a cell surface molecule such as, e.g., CD40 or CD40L (CD 154) and a second biologically active compound is a pharmaceutically active compound such as an anti-sense nucleic acid or a cytostatic compound.
  • therapeutic use would be a tumor specific antibody as first biological active compound and the second biological active compound comprises pseudomonas exotoxin, diphtheria toxin, transcription factors activating p53 production or other apoptosis inducing factors.
  • Another combination of biological active compounds could be the association of gpl20-HIV binding domain of CD4 and any antiviral or cytotoxic drug capable of blocking viral maturation or killing the infected cell.
  • bifunctional but multifunctional oligomers could be created by the invention using as one compound (not necessarily biologically active) a multivalent system, allowing the covalent association via polyionic interactions and a disulfide bridge of different biological active compounds.
  • a virus shell displaying several polyionic peptide sequences on the surface could provide such a multivalent matrix.
  • nucleic acids encoding said two polypeptide chains which are each linked to a biologically active polypeptide are expressed in a prokaryotic or eukaryotic host cell simultaneously or separately, the polypeptides are recovered from the host cell or the supernatant, treated with an oxidizing agent to form said disulfide bridges and said chimeric polypeptide is isolated.
  • Such "naturation methods" are described, for example, in US Patent 4,933,434, pages 453, 363, and US Patent 5,593,865.
  • the first and second polypeptide chains are coupled via ionic interaction at a neutral or weak basic pH (preferably pH 7 to 8.5) and at low ionic strength (preferably 0 to 200 mmol/1 NaCl).
  • the polypeptide chains are either directly covalently linked via the disulfide bridge, whereby a mixed disulfide is formed and the two polypeptide chains are linked together via disulfide bridging under oxidizing or weakly reducing conditions.
  • GSH is used in combination with GSSG whereby the ratio GSH : GSSG is from 5 : 1 to 1 : 5, at a neutral or weak basic pH value.
  • Figure 1 Formation of the disulfide bonded heterodimer ACE8-ACK8 in dependence on the NaCl concentration in the buffer.
  • the relative amounts of ACK8 (A), the mixed disulfide between ACK8 and GSSG (ACK8-SG, H ) and the disulfide linked heterodimer ACE8-ACK8 ( • ) are shown.
  • Figure 2 Formation of the disulfide bonded heterodimer ACE8-ACK8 in dependence on the redox potential of the buffer. The amount of heterodimer ACE8-ACK8 ( • ) and the non- converted peptide ACK8 ( ⁇ ) are given.
  • Figure 3 Formation of the disulfide bonded heterodimer ACE8-ACK8 in dependence on a tenfold molar excess of a cysteine containing laminin peptide and ⁇ -glucosidase is analyzed, respectively. The competition is performed in buffers with two different redox systems.
  • Figure 4 Coomassie-stained SDS-PAGE ( 18%) under reducing conditions for evidence of directed associaton of VLP's with ds Fv's; Lanes : ( 1) dsFv dissociated in VH and VL; (2) association reaction between wildtype VLP's and ds Fv in the presence of 200 mM NaCl; (3) association reaction between VLP's build up by VPl-Glu and ds Fv in the presence of 750 mM ammonium sulphate; (4) association reaction between VLP's build up by VPl-Glu and ds Fv in the presence of 200 mM NaCl; (5) molecular weight marker
  • Figure 5 Elution profile of the association reaction of FabDlOSCP and ⁇ - glucosidase R10CGP at low ionic strength (TosoHaas TSK 2000 SWXL; 50 mM Na 2 HPO 4 /NaH 2 PO 4 pH 7.0; 300 mM NaCl; flow rate 0.75 ml/min; column volume: 14.335 ml).
  • the results of the bifunctionality assay (modified ELISA) that detect only molecules containing both th Fab and the -glucosidase are shown.
  • 200 ⁇ M ACK8 was converted to the mixed disulfide form ACK8-SG using 10 mM GSSG in a buffer 500 mM sodium borate pH 8.5.
  • This mixed disulfide was purified by ion exchange chromatography.
  • the specific association and redox reaction of 20 ⁇ M ACE8 and the mixed disulfide ACK8-SG was carried out in 20 mM sodium borate pH 8.5, 2 mM EDTA, 25°C in the presence of NaCl at concentration between 0 and 1 M. After an incubation of 30 min further redox reaction was blocked by adding 20 mM iodoacetamide. Analyses of formation of heterodimers were performed by cation exchange chromatography as described before.
  • the disulfide bonded heterodimer ACK8-ACE8 was quantitatively formed at NaCl concentrations below 200 mM. At higher salt concentrations the polyionic interaction between the peptides was suppressed, leading to lower yields of heterodimer formation.
  • ACK8 and 75 ⁇ M ACE8 were incubated in 100 mM sodium phosphate pH 8.5, 2 mM EDTA at 25°C in the presence of 2.5 mM redox substances (GSH and GSSG).
  • the redox potential of the buffer was varied by changing the ratio of GSH and GSSG. After 5 h incubation the reaction was stopped by adding 100 mM iodoacetamide and analyzed as described before.
  • ACK8 in the controls was set to 100 % and the yield of heterodimer formation in the competition experiments analyzed.
  • ⁇ -glucosidase (68.1 kDa) was used as a second competitor ⁇ -glucosidase (68.1 kDa).
  • This protein contains 5 cysteines, accessible for low molecular weight thiol reagents.
  • 25 ⁇ M ACK8 and 37.5 ⁇ M ACE8 were incubated in sodium borate pH 8.5, 2 mM EDTA in the presence of 60 ⁇ M ⁇ -glucosidase.
  • the two different redox conditions were identical to those described before. Analyses were performed by RP-HPLC.
  • ACE8-ACK8 The formation of heterodimeric and covalent linked ACE8-ACK8 was not influenced by the addition of an excess of laminin peptide and ⁇ -glucosidase, respectively. Based on the polyionic interactions between ACE8 and ACK8 the dimerisation of these peptides to ACE8-ACK8 is highly specific.
  • the antigen binding activity of the Fab-fragment of MAb33 is combined with the enzymatic activity of ⁇ -glucosidase using polyionic fusion peptides leading to the formation of a bifunctional antibody derivative (chimeric polypeptide).
  • the Fab fragment of MAb 33 was modified genetically so that it contains a negatively charged fusionpeptide with an additionally cysteine residue at its C- terminus: AspAspAsp-AspAspAspAspAspAspAspSerCysPro (abbreviated as
  • the second polypeptide chain is a derivative of ⁇ - glucosidase PI from Saccharomyces cerevisiae carrying a positively charged C- terminal fusion peptide (ArgArgArgArgArgArgArgArgArgArgArgArgArgArgArgArgArgArgArgArgArgCysGlyPro (abbreviated as R i0 CGP, SEQ ID NO:5).
  • the formation of the chimeric protein includes the following steps:
  • Fab fragment of mAb (monoclonal antibody) 33 was used as one part of the chimeric protein.
  • MAb 33 is a murine antibody of subclass fclgGl directed against the dimeric muscle-specific human creatine kinase (CK-MM E. C. 2.7.3.2.) (Buckel et al., Gene 51 ( 1987) 13).
  • the Fab fragment of mAb 33 contains a disulfide bond between the light chain k (25 kD) and the heavy chain fd (25 kD).
  • the light chain of mAb 33 was encoded on plasmid pBTl 11 which is a derivative of the plasmid pBR223-3.
  • sequence and cloning strategy is described in
  • EP 0 364 926 Bl For expression the plasmid was transformed into E.coli host cells containing the plasmid pUBS520 (Brinkmann et al., Gene 85 ( 1989) 109-114). a2) Construction of plasmids encoding a fusion protein of the heavy chain with a C-terminal polyionic peptide sequence
  • Vector construction started with a plasmid pi 2016 encoding the heavy chain of mAb 33 (Buckel et al., Gene 51 ( 1987) 13).
  • the nucleotide sequences encoding the Ch2 and Ch3 domains of the heavy chain were deleted and a nucleotide sequence coding a polyionic peptide with a cysteine at its C-terminus was added at the chl domain using primer mutagenesis.
  • the nucleotide sequence contains auxiliary codons at its 5'-terminus encoding the five N-terminal amino acid residues of ⁇ - galactosidase (ITNSR) in order to facilitate protein expression in E. coli.
  • INSR ⁇ - galactosidase
  • the cDNA encoding the fd-fr agment was amplified with the primers 1 and 2 by PCR. At the 5'- terminus a Ndel restriction site was inserted. At the 3 '-terminus a Hind III restriction site and the nucletide sequence of the polyionic peptide with the additional cysteine were inserted.
  • the PCR was carried out with the following primers:
  • Reverse primer of the fdDi oSCP-variant (SEQ ID NO: 7): 5'- CAT AGT CCC AAG CTT TTA CGG GCA AGA ATC ATC GTC ATC ATC GTC GTC ATC ATC ACC ACA ATC CCT GGG CAC AAT - 3'
  • the modified cDNA-fragment was cloned into the vector pET-l la (Novagen) which belongs to the T7-expression systems (Studier, F.W., and Moffatt, B.A., J. Mol. Biol. 189 ( 1986) 113).
  • the vector was transformed into host cells containing plasmid pUBS520.
  • the plasmid pUBS520 (Brinkmann et al., Gene 85 ( 1989) 109-114) encodes a tRNA which is necessary for translation of the codons
  • the protein aggregates were solubilized in 6 M guanidine hydrochloride ( 100 mM TRIS-HCL pH 8.5; 1 mM EDTA; 100 mM DTT) at 4°C overnight.
  • the pH was reduced to 4.0 using 0.5 M HC1 and unsolubilized material was separated by centrifugation (20,000 rpm; 30 min; 4°C).
  • the solution was extensively dialysed against 4 M guanidine hydrochloride pH 4.0 in order to remove dithiothreitol.
  • the protein concentration was determined spectrophotometrically using authentic denatured, reduced Fab fragment as a standard.
  • the denatured protein was diluted 100- fold in renaturation buffer (1 M Tris/HCl pH 8.0; 2 mM EDTA; 2.4 mM GSSG/0.6 mM GSH) to a final protein concentration of 10 ⁇ g/ml. Renaturation was performed at a scale of 10 liters over 150 h at 15°C.
  • the functionality of the renatured Fab fragments was assayed by ELISA according to Buchner, J., and Rudolph, R., Bio/Technology 9 (1991) 157.
  • the renatured protein solution was centrifuged ( 13,000 rpm; 30 min; 4°C) in order to remove higher molecular aggregates and the supernatant was concentrated by cross flow filtration (tangential flow ultrafiltration; ProVario-3-System; filter cassette:
  • the retentate of the Fab fragment with the polyaspartate fusion peptide (abbreviated as FabDioSCP) was dialysed against 20 mM Tris/HCl pH 8.0 .
  • the Fab fragment FabDioSCP was purified by anion exchange chromatography using a Resource Q column (Pharmacia; column volume: 6 ml). Elution was performed in a linear sodium chloride gradient from 0 to 1 M in 20 mM Tris-HCl pH 8.0 over 20 column volumes at a flow rate of 6 ml/ min. FabDioSCP eluted at a NaCl concentration of 300 mM. The dimer was effectively separated from higher molecular, disulfide bridged fdDioSCP-chains which eluted at a sodium chloride concentration of 400 mM. II. Production of ⁇ -glucosidase with a C-terminal polyionic peptide sequence
  • Wildtype ⁇ -glucosidase from Saccharomyces cerevisiae is a monomeric protein with a molecular weight of 68 kDa. It contains five cysteines which are not involved in disulfide bonds.
  • a fusion protein was genetically constructed consisting of ⁇ -glucosidase PI and a C-terminal deca-arginine fusion peptide with an additional cysteine, glycine and proline.
  • the expression vector is a derivative of pKK223-3 (Brosius, J., and Holy, A., Proc. Natl. Acad. Sci. USA 81 ( 1984) 6929) which contains a tac-promotor and a ⁇ -lactamase gene.
  • the vector was modified so that it contains a single EcoRI restriction site at position 1600 of the ⁇ -glucosidase gene.
  • the fusion peptide encoding ten arginine residues, a cysteine, a glycine and a proline residue was inserted at the C-terminus by primer mutagenesis using standard recombinant DNA techniques.
  • the PCR was performed using the following primer sequences:
  • lactose ⁇ -glucosidase In combination with the limited induction in the presence of 0.5% lactose ⁇ -glucosidase predominantly accumulated in soluble form in the cytosol.
  • the supernatant of the crude extract was purified by cation exchange chromatography on a Resource S column (Pharmacia; 6 ml). Protein fraction containing ⁇ -gucosidase activity eluted at a NaCl concentration of 350 mM in a linear gradient of 0 to 500 mM NaCl (buffer: 10 mM K 2 HPO 4 /KH 2 PO 4 pH 6.8; 10 mM EDTA) over 20 column volumes at a flow rate of 6 ml/min.
  • Enzymatic activity was determined spectrophotometrically according to Kopetzki et al., Yeast 5 ( 1989) 11, at 405 nm and 30°C with 2 mM para-nitrophenylglucopyranoside (PNPG) (Sigma) in 100 mM K 2 HPO 4 /KH 2 PO 4 pH 6.8 as artificial substrate (Kopetzki et al., Yeast 5 (1989) 11) .
  • PNPG para-nitrophenylglucopyranoside
  • the polypeptides were incubated in equimolar amounts (3 ⁇ mol/1) at 20°C for 48 h.
  • the reaction was stopped with iodoacetamide (final concentration 20 mM in Tris-HCl pH 8.0) and the samples were separated on a 12% SDS-PAGE under oxidizing and reducing conditions. Lanes containing Fab were detected by immunobloting on nitrocellulose.
  • reaction products were separated on a gelfiltration column (TSKgel 2000 SWXL; TosoHaas) in a 50 mM Na 2 HPO 4 /NaH 2 PO 4 buffer containing 300 mM NaCl at a flow rate of 0.75 ml/min using a Vision Workstation (BioCad Vision station; Perseptive Biosystems).
  • VLP virus like particles
  • VLP's of polyoma coat-protein VP1 and disulfide bridged Fv-fragments (ds Fv) of mAb B3, based on engineered polyionic peptide sequences is another example for the present invention.
  • the invention includes the production of VLP's, the production of ds Fv-fragments and the following association. I) Production of VLP's
  • the polyoma coat protein VPl is capable of assembling in vitro into icoasaedric VLP's (Salunke, D.M., et al. Cell 46 ( 1986) 895-904; Salunke, D.M., et al., Biophysical J. 56 ( 1989) 887-904).
  • the plasmid pALVPlTAC (Leavitt, A.D., et al., J. Biol. Chem. 260 ( 1985) 12803-12809) encodes the wildtype protein and allows the recombinant production of soluble pentameric protein in E.coli.
  • the expression strain was cultivated on mineral salt medium at 30°C at a scale of 5 liters in a Biostat-Fermenter (Braun) using the fed-batch technique.
  • Six hours after induction the cells were harvested by centrifugation (8000 g, 15 min) and stored at -70°C.
  • mutant VPl-Glu 50 g cells were resuspended in 500 ml buffer
  • A 50 mM Tris HCl; pH 7.4; 5% glycerol; 2 mM EDTA; 200 mM NaCl; 4 mM DTT.
  • Cell lysis was performed by high pressure dispersion (Gaulin, 1200 bar) in presence of 1 unit/ml Benzonase, 20 ⁇ g/ml RNase and 4 tablets of complete protease inhibitor coctail (Roche Diagnostics GmbH, DE). The lysate was centifuged for 30 min at 47,000 g.
  • the first purification and concentration step consists of a fractionated ammonium sulphate precipitation between 17.5% and 27.5% saturation of salt.
  • the resuspended protein was loaded on an anion exchange column (Poros 20 HQ). In a linear gradient ranging from 200 mM to 1M NaCl in buffer A about 30 column volumes VPl-Glu eluted at 500 mM NaCl as almost homogeneous protein.
  • the purified pentameric VPl-Glu was dialyzed against buffer B (20 mM Tris, pH 7.4; 0.75 M ammonium sulphate; 5% glycerol; 1 mM CaCl2) for 2 day at 15°C. Under these conditions the formation of VLP's is induced. In order to remove the ammonium sulphate the solution of VLP's was dialyzed against buffer C (20 mM Tris, pH 7.4; 200 mM NaCl; 5 % glycerin; ImM CaCl2) for 1 day at 15°C and afterwards stored at 4°C or -20°C. II ) Production of ds Fv
  • Fragment with a polyionic fusion peptide was constructed. For this purpose a stop codon was introduced between the coding sequence of VL and the toxin part, thus creating an expression vector for the VL domain of mAb B3.
  • the V H domain, encoded on the plasmid pYR 38-2 was extended by a polyionic sequence ArgArgArgArgArgArgArgArgArgArgCysPro (R 8 CP, SEQ ID NO:l l) at the C- terminus. This extension was achieved by a two-step procedure using the QuickChange site directed mutagenenesis kit (Stratagene). First a oligonucleotide coding ArgArgArgArgCysPro was inserted at the 3'end of the VH gene (Reiter, Y., et al., Protein Engng. 12 ( 1995) 1323-1331).
  • VH- and V L -domain were separately expressed in E. coli as inclusion bodies.
  • the preparation of ib's was performed according the method by Rudolph et al. (Rudolph et al., Folding Proteins, In: T.E. Creighton (ed.): Protein function: A
  • the polycationic sequence at the C-terminus of the V ⁇ -domain allowed a purification of folded ds Fv by cation exchange chromatography.
  • the renatured protein was loaded on a Poros20 HS column and eluted with a linear gradient from
  • ds Fv elutes as a homogeneous protein at 400 mM NaCl.
  • the homogeneity of ds Fv was shown by gel filtration (Pharmacia Superdex 75), running with buffer D and SDS PAGE.
  • Tris Tris; pH 7.4; 5 % glycerol; 1 mM CaCl 2 ; 37.5 ⁇ M GSSG).
  • concentration of VPl-Glu used was 5 ⁇ M and the concentration of ds Fv 2.5 ⁇ M.
  • the reaction was performed in the presence of 0.2 M NaCl and 0.75 M ammonium sulphate, respectively.
  • wildtype VPl without polyionic sequence was mixed with ds Fv in the presence of 0.2 M NaCl.
  • the reaction mixtures were incubated for 8 h at 20°C and then applied on a gel filtration column (TosoHAAS TSK-Gel PW 6000 XL), equilibrated in buffer E and 0.2 M NaCl. Fractions containing VLP's were precipitated with sodium desoxycholate and analyzed by 18% SDS PAGE (Fig. 4) and Western Blot.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • General Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
PCT/EP2000/007378 1999-08-02 2000-07-31 Chimeric polypeptides, method for production and uses thereof WO2001009193A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
DE60033405T DE60033405T2 (de) 1999-08-02 2000-07-31 Chimäre polypeptide, verfahren zur herstellung und verwendung dafür
MXPA02001139A MXPA02001139A (es) 1999-08-02 2000-07-31 Polipeptidos quimericos, metodos para su produccion y usos de los mismos.
BR0012926-7A BR0012926A (pt) 1999-08-02 2000-07-31 Polipeptìdeos quiméricos, método para a sua produção e seus usos
AU61610/00A AU768645B2 (en) 1999-08-02 2000-07-31 Chimeric polypeptides, method for production and uses thereof
DK00948017T DK1206495T3 (da) 1999-08-02 2000-07-31 Kimære polypeptider, fremgangsmåde til frembringelse og anvendelser deraf
CA002380569A CA2380569C (en) 1999-08-02 2000-07-31 Chimeric polypeptides, method for production and uses thereof
EP00948017A EP1206495B1 (en) 1999-08-02 2000-07-31 Chimeric polypeptides, method for production and uses thereof
JP2001513999A JP4011914B2 (ja) 1999-08-02 2000-07-31 キメラポリペプチド、その製造方法、およびその使用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP99115022A EP1074563A1 (en) 1999-08-02 1999-08-02 Chimeric polypeptides enhancing dimer formation through electrostatic interactions and disulfide bond, method for production and uses thereof
EP99115022.8 1999-08-02

Publications (1)

Publication Number Publication Date
WO2001009193A1 true WO2001009193A1 (en) 2001-02-08

Family

ID=8238697

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2000/007378 WO2001009193A1 (en) 1999-08-02 2000-07-31 Chimeric polypeptides, method for production and uses thereof

Country Status (17)

Country Link
US (1) US6437095B1 (ko)
EP (2) EP1074563A1 (ko)
JP (1) JP4011914B2 (ko)
KR (1) KR100484084B1 (ko)
CN (1) CN1184235C (ko)
AR (1) AR021448A1 (ko)
AT (1) ATE353921T1 (ko)
AU (1) AU768645B2 (ko)
BR (1) BR0012926A (ko)
CA (1) CA2380569C (ko)
DE (1) DE60033405T2 (ko)
DK (1) DK1206495T3 (ko)
ES (1) ES2280231T3 (ko)
MX (1) MXPA02001139A (ko)
TR (1) TR200200252T2 (ko)
WO (1) WO2001009193A1 (ko)
ZA (1) ZA200109746B (ko)

Families Citing this family (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7192596B2 (en) * 1996-08-23 2007-03-20 The Health Protection Agency Ipsen Limited Recombinant toxin fragments
US20030157091A1 (en) * 2002-02-14 2003-08-21 Dyax Corporation Multi-functional proteins
US7335361B2 (en) * 2003-06-09 2008-02-26 Animal Technology Institute Taiwan Fusion antigen used as vaccine
US7348399B2 (en) 2003-08-29 2008-03-25 Louisiana Tech University Foundation, Inc. Nanofabricated polypeptide multilayer films, coatings, and microcapsules
US7544770B2 (en) * 2003-08-29 2009-06-09 Louisiana Tech Foundation, Inc. Multilayer films, coatings, and microcapsules comprising polypeptides
US7550557B2 (en) 2003-08-29 2009-06-23 Louisiana Tech University Foundation, Inc. Multilayer films, coatings, and microcapsules comprising polypeptides
CA2587643C (en) * 2004-11-22 2013-09-24 Louisiana Tech University Foundation Method for designing polypeptides for the nanofabrication of thin films, coatings, and microcapsules by electrostatic layer-by-layer self assembly
US20090246127A1 (en) * 2004-12-17 2009-10-01 Koninklijke Philips Electronics, N.V. Targeting agents for molecular imaging
CN104945509A (zh) 2009-09-16 2015-09-30 弗·哈夫曼-拉罗切有限公司 包含卷曲螺旋和/或系链的蛋白质复合体及其用途
TW201138821A (en) 2010-03-26 2011-11-16 Roche Glycart Ag Bispecific antibodies
CN103384831B (zh) 2010-12-23 2016-02-10 霍夫曼-拉罗奇有限公司 通过二价结合剂来检测多肽二聚体
WO2012085064A1 (en) 2010-12-23 2012-06-28 Roche Diagnostics Gmbh Detection of a posttranslationally modified polypeptide by a bi-valent binding agent
CN103384681B (zh) 2010-12-23 2018-05-18 霍夫曼-拉罗奇有限公司 结合剂
EP2655413B1 (en) 2010-12-23 2019-01-16 F.Hoffmann-La Roche Ag Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
WO2012107932A1 (en) * 2011-02-11 2012-08-16 Transgene Biotek Ltd. B cell clonal immunotoxin and use thereof
WO2012171996A1 (en) 2011-06-15 2012-12-20 F. Hoffmann-La Roche Ag Anti-human epo receptor antibodies and methods of use
WO2013119966A2 (en) 2012-02-10 2013-08-15 Genentech, Inc. Single-chain antibodies and other heteromultimers
JP6203838B2 (ja) 2012-06-27 2017-09-27 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 少なくとも2つの異なる結合実体を含む、テーラーメイドの高度に選択的かつ多重特異的なターゲティング実体を選択および作製するための方法、ならびにその使用
WO2014001325A1 (en) 2012-06-27 2014-01-03 F. Hoffmann-La Roche Ag Method for making antibody fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
CA2871386A1 (en) 2012-06-27 2014-01-03 F. Hoffmann-La Roche Ag Method for the selection and production of tailor-made, selective and multi-specific therapeutic molecules comprising at least two different targeting entities and uses thereof
MX371403B (es) 2014-03-20 2020-01-29 Bristol Myers Squibb Co Moleculas de andamiaje a base de fibronectina estabilizada.
ES2755395T3 (es) 2014-06-06 2020-04-22 Bristol Myers Squibb Co Anticuerpos contra el receptor del factor de necrosis tumoral inducido por glucocorticoides (GITR) y usos de los mismos
US10100129B2 (en) 2014-11-21 2018-10-16 Bristol-Myers Squibb Company Antibodies against CD73 and uses thereof
JP6622814B2 (ja) 2014-11-25 2019-12-18 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company イメージングのための新規pd−l1結合ポリペプチド
PL3227332T3 (pl) 2014-12-03 2020-06-15 F. Hoffmann-La Roche Ag Wielospecyficzne przeciwciała
EP3945096A1 (en) 2014-12-19 2022-02-02 Regenesance B.V. Antibodies that bind human c6 and uses thereof
WO2016127052A1 (en) 2015-02-05 2016-08-11 Bristol-Myers Squibb Company Cxcl11 and smica as predictive biomarkers for efficacy of anti-ctla4 immunotherapy
SG10202008304TA (en) 2015-05-29 2020-10-29 Bristol Myers Squibb Co Antibodies against ox40 and uses thereof
CN108884147B (zh) 2015-09-23 2024-02-27 百时美施贵宝公司 结合磷脂酰肌醇蛋白聚糖3的基于纤连蛋白的支架分子
SG11201803817PA (en) 2015-11-19 2018-06-28 Bristol Myers Squibb Co Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
BR112018067368A2 (pt) 2016-03-04 2019-01-15 Bristol-Myers Squibb Company terapia de combinação com anticorpos anti-cd73
EP3463486A1 (en) 2016-06-01 2019-04-10 Bristol-Myers Squibb Company Pet imaging with pd-l1 binding polypeptides
MY200602A (en) 2016-07-14 2024-01-04 Bristol Myers Squibb Co Antibodies against tim3 and uses thereof
WO2018083538A1 (en) 2016-11-07 2018-05-11 Neuracle Scienc3 Co., Ltd. Anti-family with sequence similarity 19, member a5 antibodies and method of use thereof
TWI788321B (zh) * 2017-01-20 2023-01-01 美商健臻公司 骨靶向抗體
CA3051839A1 (en) 2017-02-17 2018-08-23 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
CN111886255A (zh) 2018-01-12 2020-11-03 百时美施贵宝公司 抗tim3抗体及其用途
US11242393B2 (en) 2018-03-23 2022-02-08 Bristol-Myers Squibb Company Antibodies against MICA and/or MICB and uses thereof
TWI790370B (zh) 2018-04-02 2023-01-21 美商必治妥美雅史谷比公司 抗trem-1抗體及其用途
WO2020003210A1 (en) 2018-06-29 2020-01-02 Kangwon National University University-Industry Cooperation Foundation Anti-l1cam antibodies and uses thereof
BR112021014106A2 (pt) 2019-01-22 2021-10-13 Bristol-Myers Squibb Company Anticorpos contra subunidade alfa de il-7r e usos dos mesmos
CN114805598A (zh) * 2019-04-28 2022-07-29 广州市雷德生物科技有限公司 促进蛋白二聚体形成的拉链扣结构及其应用
US20220372139A1 (en) 2019-07-15 2022-11-24 Bristol-Myers Squibb Company Anti-trem-1 antibodies and uses thereof
WO2021011681A1 (en) 2019-07-15 2021-01-21 Bristol-Myers Squibb Company Antibodies against human trem-1 and uses thereof
CN112694527B (zh) * 2020-12-25 2022-09-09 山东睿鹰制药集团有限公司 一种重组人干扰素-κ包涵体的纯化和复性方法
JP2024514530A (ja) 2021-04-02 2024-04-02 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 切断型cdcp1に対する抗体およびその使用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994029350A2 (en) * 1993-06-14 1994-12-22 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Recombinant disulfide-stabilized polypeptide fragments having binding specificity
WO1998050431A2 (en) * 1997-05-02 1998-11-12 Genentech, Inc. A method for making multispecific antibodies having heteromultimeric and common components

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK132892D0 (da) * 1992-10-30 1992-10-30 Novo Nordisk As Proteiner

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994029350A2 (en) * 1993-06-14 1994-12-22 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Recombinant disulfide-stabilized polypeptide fragments having binding specificity
WO1998050431A2 (en) * 1997-05-02 1998-11-12 Genentech, Inc. A method for making multispecific antibodies having heteromultimeric and common components

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
MCPHEE F ET AL: "Engineering human immunodeficiency virus 1 protease heterodimers as macromolecular inhibitors of viral maturation.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCE OF THE UNITED STATES OF AMERICA, vol. 93, October 1996 (1996-10-01), pages 11477 - 81, XP002126434 *
O'SHEA E ET AL: "Peptide "velcro": design of a heterodimeric coiled coil", CURRENT BIOLOGY, vol. 3, no. 10, 1993, pages 658 - 67, XP000653001 *
WELLS J ET AL: "in vivo formation and stability of engineered disulfide bonds in subtilisin.", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 261, no. 14, 15 May 1986 (1986-05-15), pages 6564 - 70, XP002037078 *
ZHU Z ET AL: "Remodeling domain interfaces to enhance heterodimer formation.", PROTEIN SCIENCE, vol. 6, 1997, pages 781 - 8, XP002084764 *

Also Published As

Publication number Publication date
ZA200109746B (en) 2003-03-12
DK1206495T3 (da) 2007-06-11
JP4011914B2 (ja) 2007-11-21
JP2003506028A (ja) 2003-02-18
EP1074563A1 (en) 2001-02-07
CA2380569A1 (en) 2001-02-08
DE60033405D1 (de) 2007-03-29
US6437095B1 (en) 2002-08-20
AR021448A1 (es) 2002-07-17
AU6161000A (en) 2001-02-19
ES2280231T3 (es) 2007-09-16
KR20020026558A (ko) 2002-04-10
ATE353921T1 (de) 2007-03-15
TR200200252T2 (tr) 2002-05-21
EP1206495B1 (en) 2007-02-14
BR0012926A (pt) 2002-04-30
CN1358198A (zh) 2002-07-10
KR100484084B1 (ko) 2005-04-20
CN1184235C (zh) 2005-01-12
EP1206495A1 (en) 2002-05-22
AU768645B2 (en) 2003-12-18
DE60033405T2 (de) 2007-10-31
MXPA02001139A (es) 2002-08-20
CA2380569C (en) 2008-01-22

Similar Documents

Publication Publication Date Title
EP1206495B1 (en) Chimeric polypeptides, method for production and uses thereof
CA2916960C (en) High-stability t-cell receptor and preparation method and application thereof
JP2020128415A (ja) エンジニアリングされたFc構築物に関連する組成物および方法
US8383575B2 (en) (DI)barnase-barstar complexes
AU621358B2 (en) Mutant human angiogenin (angiogenesis factor with superior angiogenin activity) genes therefor and methods of expression
JP2017529059A5 (ko)
CA2754408A1 (en) Fusion proteins comprising canine fc portions
JP2013519392A (ja) Hsa関連組成物および使用方法
WO2014178078A2 (en) Novel cloning, expression & purification method for the preparation of ranibizumab
CN100357314C (zh) 一种分子物质连接的方法
EP0618227A1 (en) Biologically active polypeptide fusion dimers
US20100015123A1 (en) Novel thrombolytic molecules and a process therefor
JP2005518344A (ja) 親和性向上剤
WO2015090234A1 (en) Improving pharmacokinetic profile for angiopoietin-2 inhibiting polypeptide or thymalfasin
US20200031895A1 (en) Stabilized proteolytically activated growth differentiation factor 11
CN107223133B (zh) 一种可溶的异质二聚t细胞受体及其制法和应用
US11028132B1 (en) Half-life optimized linker composition
CN114716563B (zh) 一种融合蛋白及其制备与应用
CN115819611A (zh) 生长激素融合蛋白及其制备方法和用途
Yan et al. Expression, refolding, and characterization of GFE peptide-fused human interferon-α2a in Escherichia coli
JPWO2021083077A5 (ko)
US20070167610A1 (en) Recombinant soluble adenovirus receptor
AU2013211503A1 (en) FGF21 mutants and uses thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 00809515.9

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2001/09746

Country of ref document: ZA

Ref document number: 200109746

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 61610/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2002/00252

Country of ref document: TR

WWE Wipo information: entry into national phase

Ref document number: 2380569

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: PA/a/2002/001139

Country of ref document: MX

Ref document number: 2000948017

Country of ref document: EP

Ref document number: 1020027001377

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 1020027001377

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2000948017

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 1020027001377

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 2000948017

Country of ref document: EP